1
|
Kim K, Kim JH, Kim I, Seong S, Kook H, Koh JT, Kim N. Tripartite motif-containing 27 negatively regulates NF-κB activation in bone remodeling. Mol Med 2025; 31:141. [PMID: 40251491 PMCID: PMC12008848 DOI: 10.1186/s10020-025-01204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Tripartite motif-containing 27 (TRIM27) is highly expressed in the mouse thymus, spleen, and hematopoietic compartment cells and regulates cell proliferation, apoptosis, and innate immune responses. However, the role of TRIM27 in bone remodeling remains unknown. This study aimed to investigate the role of TRIM27 in the differentiation of osteoclasts and osteoblasts. METHODS We measured the effects of overexpression or knockdown of TRIM27 in osteoclasts and osteoblasts using real-time PCR and Western blot analysis to quantify the mRNA and protein levels of marker genes. Additionally, we performed an in vivo analysis of TRIM27 knockout mice through bone mineral density analysis and histological analysis. RESULTS TRIM27 deficiency decreased bone mineral density by enhancing osteoclast differentiation and inhibiting osteoblast differentiation. Overexpression of TRIM27 in osteoclast precursors suppressed osteoclast formation and resorption activity, and ectopic expression of TRIM27 in osteoblast precursors induced osteoblast differentiation and mineralization. Additionally, we found that TRIM27 attenuated NF-κB activation in both osteoclasts and osteoblasts by interacting with TAB2 and promoting TAB2 degradation through lysosomal-dependent pathways, thereby inhibiting NF-κB signaling. CONCLUSIONS Our results identify TRIM27 as a novel negative regulator of NF-κB in bone remodeling, suggesting that regulating TRIM27 may be useful in developing treatments for musculoskeletal diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
2
|
Luo B, Liang Z, Lin W, Li Y, Zhong W, Bai D, Hu X, Xie J, Li X, Wang P, Zhu X, Zhang R, Yang L. Aqueous extract of Rehmanniae Radix Praeparata improves bone health in ovariectomized rats by modulating the miR-29a-3p/NFIA/Wnt signaling pathway axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119549. [PMID: 40024453 DOI: 10.1016/j.jep.2025.119549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/23/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rehmanniae Radix Praeparata (RRP), a widely used traditional Chinese medicine and a processed form of Rehmannia glutinosa, is primarily utilized to supplement kidney function and promote bone health. Clinical evidence suggests that RRP exhibits significant efficacy in the treatment of osteoporosis (OP). However, the precise mechanisms underlying its therapeutic effects remain incompletely understood. AIM OF THE STUDY OP is a systemic skeletal disorder characterized by reduced bone density and quality, leading to an increased risk of fractures. The aim of this study is to evaluate the effectiveness and underlying mechanisms of RRP in treating OP. MATERIALS AND METHODS Ovariectomized (OVX) rats were administered RRP aqueous extract via gavage for three months. After the treatment period, femoral microstructure and osteogenic protein levels were assessed to evaluate the efficacy of RRP. Serum exosomes (Exos) derived from different groups of rats were isolated and characterized. The levels of miR-29a-3p in serum-derived Exos and femoral tissue were quantified. Subsequently, Exos were co-cultured with rat bone marrow mesenchymal stem cells (rBMSCs) to investigate their role in promoting osteogenic differentiation and explore the molecular mechanisms underlying this process, particularly through the miR-29a-3p/NFIA/Wnt signaling pathway axis. RESULTS OVX rats exhibited significant bone microdamage. In contrast, the RRP-treated OVX rats showed marked improvements in femoral bone microstructure and increased osteogenic protein expression. MiR-29a-3p levels were elevated in serum-derived Exos from the RRP-treated rats. Furthermore, rBMSCs treated with these Exos displayed an increase in miR-29a-3p expression. Further investigations revealed that miR-29a-3p promoted osteogenesis by inhibiting NFIA expression in both bone tissue and rBMSCs. Overexpression of NFIA reversed the osteogenic effects of miR-29a-3p, confirming NFIA as its direct target and suggesting that miR-29a-3p enhances osteogenesis by inhibiting NFIA. Additionally, NFIA was found to promote the transcription of SFRP1, an inhibitor of the Wnt signaling pathway. Our findings suggest that the RRP aqueous extract increases miR-29a-3p levels in serum Exos, which in turn inhibits NFIA and activates the Wnt signaling pathway, thereby promoting osteogenesis. CONCLUSION These findings suggest that the RRP aqueous extract improves bone health and mitigates bone microstructural damage caused by OP through the regulation of the miR-29a-3p/NFIA/Wnt signaling pathway axis.
Collapse
Affiliation(s)
- Bingjie Luo
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Ziwen Liang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Weiwen Lin
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Wenqiang Zhong
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Donghui Bai
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Xueling Hu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Ji Xie
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Xiaoyun Li
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Panpan Wang
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Xiaofeng Zhu
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510630, PR China
| | - Ronghua Zhang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China.
| | - Li Yang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong, 510630, PR China.
| |
Collapse
|
3
|
Zheng L, Lan C, Gao X, Zhu A, Chen Y, Lin J, Yan S, Shen X. Landscape analysis of m6A modification reveals the dysfunction of bone metabolism in osteoporosis mice. Heliyon 2025; 11:e42123. [PMID: 39991256 PMCID: PMC11847259 DOI: 10.1016/j.heliyon.2025.e42123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/19/2025] [Accepted: 01/19/2025] [Indexed: 02/25/2025] Open
Abstract
Osteoporosis (OP) is a prevalent chronic bone metabolic disorder that affects the elderly population, leading to an increased susceptibility to bone fragility. Despite extensive research on the onset and progression of OP, the precise mechanisms underlying this condition remain elusive. The m6A modification, a prevalent form of chemical RNA modification, primarily regulates posttranscriptional processes, including RNA stability, splicing, and translation. Numerous studies have underscored the crucial functions of m6A regulators in OP. This study aimed to explore the relationship between OP and RNA m6A methylation, investigating its underlying mechanisms through comprehensive bioinformatic analysis and experimental validation. The mRNA sequencing (mRNA-seq) and methylated RNA immunoprecipitation sequencing (MeRIP-seq) were performed on control mice as well as ovariectomized mice to discover differentially expressed genes (DEGs) and m6A regulators in OP. The results revealed dysregulation of a majority of bone metabolism-related genes and m6A regulators in ovariectomized mice, indicating a closely linked relationship between them. Our research findings indicated that m6A modification is essential in regulating OP, offering potential insights for prevention and treatment.
Collapse
Affiliation(s)
- Lifeng Zheng
- Department of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Orthopedics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Chao Lan
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Xinyue Gao
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, 350108, China
| | - An Zhu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, 350108, China
| | - Yaoqing Chen
- Department of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Orthopedics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jinluan Lin
- Department of Orthopedics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Orthopedics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Sunjie Yan
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Ximei Shen
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| |
Collapse
|
4
|
Zhou R, Wang J. Identification of Metabolism-Related Prognostic Biomarkers and Immune Features of Head and Neck Squamous Cell Carcinoma. Crit Rev Immunol 2024; 44:61-78. [PMID: 38505922 DOI: 10.1615/critrevimmunol.2024050754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
We aimed to identify an effective metabolic subtype and risk score to predict survival and immunotherapy response in head and neck squamous cell carcinoma (HNSCC). Data were obtained from an online database. We screened significant prognostic metabolism-related genes between the normal and tumor groups using a series of bioinformatics methods. Based on the selected prognostic genes, we conducted a subtype analysis to identify significantly different subtypes in HNSCC. We then investigated survival, immune features, and hallmark differences among different subtypes. LASSO was utilized to identify optimal genes for the risk score model construction. Finally, distribution of the risk score samples was analyzed for different subtypes. A total of 32 significantly prognostic metabolism-related genes were screened, and all samples were grouped into two subtypes: cluster 1 and cluster 2. Cluster 1 had worse survival. Different immune cell infiltration (CD8 T cells, macrophages, and regulatory T cells) and immune checkpoint gene expression (PD-1 and CLAT-4) were observed between the two clusters. Twelve optimal genes were involved in risk score model, and high-risk group had poorer survival. Cluster 1 contained more high-risk samples (60%). Finally, four genes CAV1, GGT6, PYGL, and HS3ST1 were identified as significantly related to immune cells, and these genes were differentially expressed in the normal oral epithelial cells and HNSCC cells. The subtypes and risk score model in the study provide a promising biomarker for prognosis and immunotherapy response.
Collapse
Affiliation(s)
- Rongjin Zhou
- Department of Ophthalmology and Otorhinolaryngology, Dongtai People's Hospital, Yancheng 224200, China
| | - Junguo Wang
- Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory)
| |
Collapse
|
5
|
Chen Y, Tang B, Jiang W, Sun M, Zhang H, Tao Y, Wang H, Xiang D, Bai H, Guo M, Zhao P, Yan W, Huang X, Chen T, Lian C, Zhang J. miR-486-5p Attenuates Steroid-Induced Adipogenesis and Osteonecrosis of the Femoral Head Via TBX2/P21 Axis. Stem Cells 2023; 41:711-723. [PMID: 37210668 DOI: 10.1093/stmcls/sxad038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023]
Abstract
Enhanced adipogenic differentiation of mesenchymal stem cells (MSCs) is considered as a major risk factor for steroid-induced osteonecrosis of the femoral head (SOFNH). The role of microRNAs during this process has sparked interest. miR-486-5p expression was down-regulated significantly in femoral head bone tissues of both SONFH patients and rat models. The purpose of this study was to reveal the role of miR-486-5p on MSCs adipogenesis and SONFH progression. The present study showed that miR-486-5p could significantly inhibit adipogenesis of 3T3-L1 cells by suppressing mitotic clonal expansion (MCE). And upregulated expression of P21, which was caused by miR-486-5p mediated TBX2 decrease, was responsible for inhibited MCE. Further, miR-486-5p was demonstrated to effectively inhibit steroid-induced fat formation in the femoral head and prevented SONFH progression in a rat model. Considering the potent effects of miR-486-5p on attenuating adipogenesis, it seems to be a promising target for the treatment of SONFH.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Boyu Tang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Weiqian Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Mingjie Sun
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hongrui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yuzhang Tao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hongwei Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dulei Xiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Haobo Bai
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Mingkang Guo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Pei Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Wenlong Yan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xiao Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Tingmei Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Chengjie Lian
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jian Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
6
|
Tozawa T, Matsunaga K, Izumi T, Shigehisa N, Uekita T, Taoka M, Ichimura T. Ubiquitination-coupled liquid phase separation regulates the accumulation of the TRIM family of ubiquitin ligases into cytoplasmic bodies. PLoS One 2022; 17:e0272700. [PMID: 35930602 PMCID: PMC9355226 DOI: 10.1371/journal.pone.0272700] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
Many members of the tripartite motif (TRIM) family of ubiquitin ligases localize in spherical, membrane-free structures collectively referred to as cytoplasmic bodies (CBs) in a concentration-dependent manner. These CBs may function as aggresome precursors or storage compartments that segregate potentially harmful excess TRIM molecules from the cytosolic milieu. However, the manner in which TRIM proteins accumulate into CBs is unclear. In the present study, using TRIM32, TRIM5α and TRIM63 as examples, we demonstrated that CBs are in a liquid droplet state, resulting from liquid-liquid phase separation (LLPS). This finding is based on criteria that defines phase-separated structures, such as recovery after photobleaching, sensitivity to hexanediol, and the ability to undergo fusion. CB droplets, which contain cyan fluorescent protein (CFP)-fused TRIM32, were purified from HEK293 cells using a fluorescence-activated cell sorter and analyzed by LC-MS/MS. We found that in addition to TRIM32, these droplets contain a variety of endogenous proteins and enzymes including ubiquitin. Localization of ubiquitin within CBs was further verified by fluorescence microscopy. We also found that the activation of the intracellular ubiquitination cascade promotes the assembly of TRIM32 molecules into CBs, whereas inhibition causes suppression. Regulation is dependent on the intrinsic E3 ligase activity of TRIM32. Similar regulation by ubiquitination on the TRIM assembly was also observed with TRIM5α and TRIM63. Our findings provide a novel mechanical basis for the organization of CBs that couples compartmentalization through LLPS with ubiquitination.
Collapse
Affiliation(s)
- Takafumi Tozawa
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Kanagawa, Japan
| | - Kohichi Matsunaga
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Tetsuro Izumi
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Naotake Shigehisa
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Kanagawa, Japan
| | - Takamasa Uekita
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Kanagawa, Japan
| | - Masato Taoka
- Department of Chemistry, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Tohru Ichimura
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
7
|
Wang XM, Zhang Y, Mannan R, Skala SL, Rangaswamy R, Chinnaiyan A, Su F, Cao X, Zelenka-Wang S, McMurry L, Xiao H, Spratt DE, Sangoi A, Shao L, Betz BL, Brown N, Tickoo SK, McKenney JK, Argani P, Gupta S, Reuter VE, Chinnaiyan AM, Dhanasekaran SM, Mehra R. TRIM63 is a sensitive and specific biomarker for MiT family aberration-associated renal cell carcinoma. Mod Pathol 2021; 34:1596-1607. [PMID: 33854184 PMCID: PMC8298271 DOI: 10.1038/s41379-021-00803-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 12/20/2022]
Abstract
Microphthalmia-associated transcription factor (MiT) family aberration-associated renal cell carcinoma (MiTF-RCC) is a subtype of renal cell carcinoma harboring recurrent chromosomal rearrangements involving TFE3 or TFEB genes. MiTF-RCC is morphologically diverse, can histologically resemble common RCC subtypes like clear cell RCC and papillary RCC, and often poses a diagnostic challenge in genitourinary clinical and pathology practice. To characterize the MiTF-RCC at the molecular level and identify biomarker signatures associated with MiTF-RCC, we analyzed RNAseq data from MiTF-RCC, other RCC subtypes and benign kidney. Upon identifying TRIM63 as a cancer-specific biomarker in MiTF-RCC, we evaluated its expression independently by RNA in situ hybridization (RNA-ISH) in whole tissue sections from 177 RCC cases. We specifically included 31 cytogenetically confirmed MiTF-RCC cases and 70 RCC cases suspicious for MiTF-RCC in terms of clinical and morphological features, to evaluate and compare TRIM63 RNA-ISH results with the results from TFE3/TFEB fluorescence in situ hybridization (FISH), which is the current clinical standard. We confirmed that TRIM63 mRNA was highly expressed in all classes of MiTF-RCC compared to other renal tumor categories, where it was mostly absent to low. While the TRIM63 RNA-ISH and TFE3/TFEB FISH results were largely concordant, importantly, TRIM63 RNA-ISH was strongly positive in TFE3 FISH false-negative cases with RBM10-TFE3 inversion. In conclusion, TRIM63 can serve as a diagnostic marker to distinguish MiTF-RCC from other renal tumor subtypes with overlapping morphology. We suggest a combination of TFE3/TFEB FISH and TRIM63 RNA-ISH assays to improve the accuracy and efficiency of MiTF-RCC diagnosis. Accurate diagnosis of MiTF-RCC and other RCC subtypes would enable effective targeted therapy and avoid poor therapeutic response due to tumor misclassification.
Collapse
Affiliation(s)
- Xiao-Ming Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI,Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Yuping Zhang
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Rahul Mannan
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Stephanie L. Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | | | | | - Fengyun Su
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Xuhong Cao
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Sylvia Zelenka-Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI,Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Lisa McMurry
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Hong Xiao
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Daniel E. Spratt
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI
| | - Ankur Sangoi
- Department of Pathology, El Camino Hospital, Mountain View, CA
| | - Lina Shao
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Bryan L. Betz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Noah Brown
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Satish K. Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jesse K. McKenney
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Anatomic Pathology, Cleveland Clinic, Cleveland, OH
| | - Pedram Argani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Victor E. Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Arul M. Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI,Michigan Center for Translational Pathology, Ann Arbor, MI,Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI,Department of Urology, University of Michigan Medical School, Ann Arbor, MI,Howard Hughes Medical Institute, Ann Arbor, MI
| | - Saravana M. Dhanasekaran
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI,Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA. .,Michigan Center for Translational Pathology, Ann Arbor, MI, USA. .,Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Mishra BH, Mishra PP, Raitoharju E, Marttila S, Mononen N, Sievänen H, Viikari J, Juonala M, Laaksonen M, Hutri-Kähönen N, Kähönen M, Raitakari OT, Lehtimäki T. Modular genome-wide gene expression architecture shared by early traits of osteoporosis and atherosclerosis in the Young Finns Study. Sci Rep 2021; 11:7111. [PMID: 33782480 PMCID: PMC8007808 DOI: 10.1038/s41598-021-86536-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
We analysed whole blood genome-wide expression data to identify gene co-expression modules shared by early traits of osteoporosis and atherosclerosis. Gene expression was profiled for the Young Finns Study participants. Bone mineral density and content were measured as early traits of osteoporosis. Carotid and bulbus intima media thickness were measured as early traits of atherosclerosis. Joint association of the modules, identified with weighted co-expression analysis, with early traits of the diseases was tested with multivariate analysis. Among the six modules significantly correlated with early traits of both the diseases, two had significant (adjusted p-values (p.adj) < 0.05) and another two had suggestively significant (p.adj < 0.25) joint association with the two diseases after adjusting for age, sex, body mass index, smoking habit, alcohol consumption, and physical activity. The three most significant member genes from the significant modules were NOSIP, GXYLT2, and TRIM63 (p.adj ≤ 0.18). Genes in the modules were enriched with biological processes that have separately been found to be involved in either bone metabolism or atherosclerosis. The gene modules and their most significant member genes identified in this study support the osteoporosis-atherosclerosis comorbidity hypothesis and can provide new joint biomarkers for both diseases and their dual prevention.
Collapse
Affiliation(s)
- Binisha H Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland.
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Saara Marttila
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Gerontology Research Center (GEREC), Tampere University, Tampere, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Harri Sievänen
- The UKK Institute for Health Promotion Research, Tampere, Finland
| | - Jorma Viikari
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Markus Juonala
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | | | - Nina Hutri-Kähönen
- Department of Paediatrics, Tampere University Hospital, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
9
|
Chen WT, Zhang F, Zhao XQ, Yu B, Wang BW. Galectin-3 and TRIM16 coregulate osteogenic differentiation of human bone marrow-derived mesenchymal stem cells at least partly via enhancing autophagy. Bone 2020; 131:115059. [PMID: 31521826 DOI: 10.1016/j.bone.2019.115059] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) is critical for bone homeostasis. Here, we investigated the regulation of Galectin-3 and tripartite motif protein 16 (TRIM16) on osteogenic differentiation of hBMSCs through autophagy. METHODS Quantitative PCR (qPCR) and western blot were performed to determine the expression of osteogenic markers, autophagic markers, Galectin-3 and TRIM16. Short-hairpin RNAs (shRNAs) and overexpression plasmids were used to manipulate the expression of Galectin-3, TRIM16 and Unc-51 like autophagy activating kinase 1 (ULK1). Alkaline phosphatase (ALP) activity was measured by ALP staining assay. Calcium deposition in differentiated hBMSCs was assessed by Alizarin Red S staining. LC3 puncta formation was monitored by immunofluorescence staining. The interaction between indicated proteins was confirmed by co-immunoprecipitation (Co-IP) assay. RESULTS Either Galectin-3 or TRIM16 knockdown led to impaired ALP activity, reduced calcium deposition, down-regulation of pro-osteogenic markers as well as restrained autophagy in osteogenic-induced hBMSCs. However, overexpression of Galectin-3 or TRIM16 promoted osteogenic differentiation of hBMSCs, which was then compromised by autophagy inhibition. Co-IP experiment demonstrated that TRIM16 associated with Galectin-3 through ULK1. Meanwhile, osteogenic induction enhanced the association between TRIM16 and ULK1 or coiled-coil myosin-like BCL2-interacting protein (Beclin1), and TRIM16 increased the stability of ULK1 and Beclin1. Moreover, either TRIM16 or ULK1 knockdown dampened the pro-osteogenic effect of Galectin-3, which elucidated that Galectin-3 mediated osteogenic differentiation was at least partly dependent on TRIM16 and ULK1. CONCLUSION In summary, the present study revealed Galectin-3 and TRIM16 co-regulated osteogenic differentiation of hBMSCs at least partly via enhancing autophagy, which might provide a promising approach for osteoporosis treatment in future.
Collapse
Affiliation(s)
- Wen-Ting Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Fan Zhang
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Xing-Qi Zhao
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Bin Yu
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| | - Bo-Wei Wang
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
10
|
The shift in the balance between osteoblastogenesis and adipogenesis of mesenchymal stem cells mediated by glucocorticoid receptor. Stem Cell Res Ther 2019; 10:377. [PMID: 31805987 PMCID: PMC6896503 DOI: 10.1186/s13287-019-1498-0] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/09/2019] [Accepted: 11/18/2019] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells capable of differentiating into several tissues, such as bone, cartilage, and fat. Glucocorticoids affect a variety of biological processes such as proliferation, differentiation, and apoptosis of various cell types, including osteoblasts, adipocytes, or chondrocytes. Glucocorticoids exert their function by binding to the glucocorticoid receptor (GR). Physiological concentrations of glucocorticoids stimulate osteoblast proliferation and promote osteogenic differentiation of MSCs. However, pharmacological concentrations of glucocorticoids can not only induce apoptosis of osteoblasts and osteocytes but can also reduce proliferation and inhibit the differentiation of osteoprogenitor cells. Several signaling pathways, including the Wnt, TGFβ/BMP superfamily and Notch signaling pathways, transcription factors, post-transcriptional regulators, and other regulators, regulate osteoblastogenesis and adipogenesis of MSCs mediated by GR. These signaling pathways target key transcription factors, such as Runx2 and TAZ for osteogenesis and PPARγ and C/EBPs for adipogenesis. Glucocorticoid-induced osteonecrosis and osteoporosis are caused by various factors including dysfunction of bone marrow MSCs. Transplantation of MSCs is valuable in regenerative medicine for the treatment of osteonecrosis of the femoral head, osteoporosis, osteogenesis imperfecta, and other skeletal disorders. However, the mechanism of inducing MSCs to differentiate toward the osteogenic lineage is the key to an efficient treatment. Thus, a better understanding of the molecular mechanisms behind the imbalance between GR-mediated osteoblastogenesis and adipogenesis of MSCs would not only help us to identify the pathogenic causes of glucocorticoid-induced osteonecrosis and osteoporosis but also promote future clinical applications for stem cell-based tissue engineering and regenerative medicine. Here, we primarily review the signaling mechanisms involved in adipogenesis and osteogenesis mediated by GR and discuss the factors that control the adipo-osteogenic balance.
Collapse
|
11
|
Magomedova L, Tiefenbach J, Zilberman E, Le Billan F, Voisin V, Saikali M, Boivin V, Robitaille M, Gueroussov S, Irimia M, Ray D, Patel R, Xu C, Jeyasuria P, Bader GD, Hughes TR, Morris QD, Scott MS, Krause H, Angers S, Blencowe BJ, Cummins CL. ARGLU1 is a transcriptional coactivator and splicing regulator important for stress hormone signaling and development. Nucleic Acids Res 2019; 47:2856-2870. [PMID: 30698747 PMCID: PMC6451108 DOI: 10.1093/nar/gkz010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 12/17/2022] Open
Abstract
Stress hormones bind and activate the glucocorticoid receptor (GR) in many tissues including the brain. We identified arginine and glutamate rich 1 (ARGLU1) in a screen for new modulators of glucocorticoid signaling in the CNS. Biochemical studies show that the glutamate rich C-terminus of ARGLU1 coactivates multiple nuclear receptors including the glucocorticoid receptor (GR) and the arginine rich N-terminus interacts with splicing factors and binds to RNA. RNA-seq of neural cells depleted of ARGLU1 revealed significant changes in the expression and alternative splicing of distinct genes involved in neurogenesis. Loss of ARGLU1 is embryonic lethal in mice, and knockdown in zebrafish causes neurodevelopmental and heart defects. Treatment with dexamethasone, a GR activator, also induces changes in the pattern of alternatively spliced genes, many of which were lost when ARGLU1 was absent. Importantly, the genes found to be alternatively spliced in response to glucocorticoid treatment were distinct from those under transcriptional control by GR, suggesting an additional mechanism of glucocorticoid action is present in neural cells. Our results thus show that ARGLU1 is a novel factor for embryonic development that modulates basal transcription and alternative splicing in neural cells with consequences for glucocorticoid signaling.
Collapse
Affiliation(s)
- Lilia Magomedova
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Jens Tiefenbach
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Emma Zilberman
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Florian Le Billan
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Veronique Voisin
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Michael Saikali
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Vincent Boivin
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Melanie Robitaille
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Serge Gueroussov
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Manuel Irimia
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Debashish Ray
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Rucha Patel
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - ChangJiang Xu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Pancharatnam Jeyasuria
- Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Gary D Bader
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Timothy R Hughes
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Quaid D Morris
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michelle S Scott
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Henry Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada.,Department of Biochemistry,University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Benjamin J Blencowe
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| |
Collapse
|
12
|
Oh SM, Shin JS, Kim IK, Kim JH, Moon JS, Lee SK, Lee JH. Therapeutic Effects of HIF-1α on Bone Formation around Implants in Diabetic Mice Using Cell-Penetrating DNA-Binding Protein. Molecules 2019; 24:molecules24040760. [PMID: 30791543 PMCID: PMC6412638 DOI: 10.3390/molecules24040760] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/15/2019] [Accepted: 02/18/2019] [Indexed: 12/21/2022] Open
Abstract
Patients with uncontrolled diabetes are susceptible to implant failure due to impaired bone metabolism. Hypoxia-inducible factor 1α (HIF-1α), a transcription factor that is up-regulated in response to reduced oxygen during bone repair, is known to mediate angiogenesis and osteogenesis. However, its function is inhibited under hyperglycemic conditions in diabetic patients. This study thus evaluates the effects of exogenous HIF-1α on bone formation around implants by applying HIF-1α to diabetic mice and normal mice via a protein transduction domain (PTD)-mediated DNA delivery system. Implants were placed in the both femurs of diabetic and normal mice. HIF-1α and placebo gels were injected to implant sites of the right and left femurs, respectively. We found that bone-to-implant contact (BIC) and bone volume (BV) were significantly greater in the HIF-1α treated group than placebo in diabetic mice (p < 0.05). Bioinformatic analysis showed that diabetic mice had 216 differentially expressed genes (DEGs) and 21 target genes. Among the target genes, NOS2, GPNMB, CCL2, CCL5, CXCL16, and TRIM63 were found to be associated with bone formation. Based on these results, we conclude that local administration of HIF-1α via PTD may boost bone formation around the implant and induce gene expression more favorable to bone formation in diabetic mice.
Collapse
Affiliation(s)
- Sang-Min Oh
- Department of Prosthodontics, College of Dentistry, Yonsei University, 134 Shinchon-dong, Seodaemoon-gu, Seoul 03722, Korea.
| | - Jin-Su Shin
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 134 Shinchon-dong, Seodaemoon-gu, Seoul 03722, Korea.
| | - Il-Koo Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 134 Shinchon-dong, Seodaemoon-gu, Seoul 03722, Korea.
| | - Jung-Ho Kim
- Research Institute for Precision Immuno-medicine, Good T Cells Incorporated, 134 Shinchon-dong, Seodaemoon-gu, Seoul 03722, Korea.
| | - Jae-Seung Moon
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 134 Shinchon-dong, Seodaemoon-gu, Seoul 03722, Korea.
| | - Sang-Kyou Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 134 Shinchon-dong, Seodaemoon-gu, Seoul 03722, Korea.
- Research Institute for Precision Immuno-medicine, Good T Cells Incorporated, 134 Shinchon-dong, Seodaemoon-gu, Seoul 03722, Korea.
| | - Jae-Hoon Lee
- Department of Prosthodontics, College of Dentistry, Yonsei University, 134 Shinchon-dong, Seodaemoon-gu, Seoul 03722, Korea.
| |
Collapse
|
13
|
Kim K, Kim JH, Kim I, Seong S, Kim N. TRIM38 regulates NF-κB activation through TAB2 degradation in osteoclast and osteoblast differentiation. Bone 2018; 113:17-28. [PMID: 29753717 DOI: 10.1016/j.bone.2018.05.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/12/2018] [Accepted: 05/09/2018] [Indexed: 11/21/2022]
Abstract
The tripartite motif protein 38 (TRIM38), a member of the TRIM family, is involved in various cellular processes such as cell proliferation, differentiation, apoptosis, and antiviral defense. However, the role of TRIM38 in osteoclast and osteoblast differentiation is not yet known. In this study, we report the involvement of TRIM38 in osteoclast and osteoblast differentiation. Overexpression of TRIM38, in osteoclast precursor cells, attenuated receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclast formation, RANKL-triggered NF-κB activation, and expression of osteoclast marker genes, such as NFATc1, osteoclast-associated receptor (OSCAR), and tartrate-resistant acid phosphatase (TRAP); and down-regulation of TRIM38 expression showed the opposite effects. Ectopic expression of TRIM38 in osteoblast precursors induced increased osteoblast differentiation and function. Elevated expression of alkaline phosphatase (ALP), bone sialoprotein (BSP), and osteocalcin was also observed due to blockade of NF-κB activation. Conversely, knockdown of TRIM38 showed the opposite effects. TRIM38 also induced degradation of lysosome-dependent transforming growth factor beta-activated kinase 1 and MAP3K7-binding protein 2 (TAB2), further blocking NF-κB activation. Taken together, our data suggest that TRIM38 plays a critical role in bone remodeling as a negative regulator of NF-κB in both osteoclast and osteoblast differentiation.
Collapse
Affiliation(s)
- Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| |
Collapse
|
14
|
Abstract
Objective: To review the recent developments in the mechanisms of epithelium sodium channels (ENaCs) induced bone formation and regulation. Data Sources: Studies written in English or Chinese were searched using Medline, PubMed and the index of Chinese-language literature with time restriction from 2005 to 2014. Keywords included ENaC, bone, bone formation, osteonecrosis, estrogen, and osteoporosis. Data from published articles about the structure of ENaC, mechanism of ENaC in bone formation in recent domestic and foreign literature were selected. Study Selection: Abstract and full text of all studies were required to obtain. Studies those were not accessible and those did not focus on the keywords were excluded. Results: ENaCs are tripolymer ion channels which are assembled from homologous α, β, and γ subunits. Crystal structure of ENaCs suggests that ENaC has a central ion-channel located in the central symmetry axis of the three subunits. ENaCs are protease sensitive channels whose iron-channel activity is regulated by the proteolytic reaction. Channel opening probability of ENaCs is regulated by proteinases, mechanical force, and shear stress. Several molecules are involved in regulation of ENaCs in bone formation, including nitride oxide synthases, voltage-sensitive calcium channels, and cyclooxygenase-2. Conclusion: The pathway of ENaC involved in shear stress has an effect on stimulating osteoblasts even bone formation by estrogen interference.
Collapse
Affiliation(s)
| | | | - Wei-Hua Xu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
15
|
Kawaguchi Y, Taoka M, Takekiyo T, Uekita T, Shoji I, Hachiya N, Ichimura T. TRIM32-Cytoplasmic-Body Formation Is an ATP-Consuming Process Stimulated by HSP70 in Cells. PLoS One 2017; 12:e0169436. [PMID: 28052117 PMCID: PMC5215751 DOI: 10.1371/journal.pone.0169436] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/05/2016] [Indexed: 12/19/2022] Open
Abstract
The spontaneous and energy-releasing reaction of protein aggregation is typically prevented by cellular quality control machinery (QC). TRIM32 is a member of the TRIM (tripartite motif-containing) ubiquitin E3 ligases, and when overexpressed in cultured cells, readily forms spherical inclusions designated as cytoplasmic bodies (CBs) even without proteasome inhibition. Here, we show that HSP70, a central QC component, is a primary binding factor of overexpressed TRIM32. Contrary to expectation, however, we find that this molecular chaperone facilitates and stabilizes CB assembly depending on intrinsic ATPase activity, rather than preventing CB formation. We also show that the HSP70-TRIM32 complex is biochemically distinct from the previously characterized 14-3-3-TRIM32 phospho-complex. Moreover, the two complexes have opposing roles, with HSP70 stimulating CB formation and 14-3-3 retaining TRIM32 in a diffuse form throughout the cytosol. Our results suggest that CB inclusion formation is actively controlled by cellular QC and requires ATP, similar to protein folding and degradation reactions.
Collapse
Affiliation(s)
- Yuki Kawaguchi
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Kanagawa, Japan
| | - Masato Taoka
- Department of Chemistry, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Takahiro Takekiyo
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Kanagawa, Japan
| | - Takamasa Uekita
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Kanagawa, Japan
| | - Ikuo Shoji
- Division of Infectious Disease Control, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naomi Hachiya
- Biotechnology Group, R&D Division, Tokyo Metropolitan Industrial Technology Research Institute, Koto-ku, Tokyo, Japan
| | - Tohru Ichimura
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
16
|
Liu Y, Wang Y, Yang N, Wu S, Lv Y, Xu L. In silico analysis of the molecular mechanism of postmenopausal osteoporosis. Mol Med Rep 2015; 12:6584-90. [PMID: 26329309 PMCID: PMC4626159 DOI: 10.3892/mmr.2015.4283] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 06/09/2015] [Indexed: 01/08/2023] Open
Abstract
Postmenopausal osteoporosis (PO) is a common disease in females >50 years of age worldwide and is becoming an increasing burden to society. The present study aimed to assess the molecular mechanism of PO using bioinformatic methods. The gene expression data from patients with PO and normal controls were downloaded from the ArrayExpress database provided by European Bioinformatics Institute. Following the screening of the differentially expressed genes (DEGs) using the Limma package in R language, Kyoto Encyclopedia of Genes and Genomes pathways enrichment analysis was performed using the Database for Annotation, Visualization and Integrated Discovery online tools. Sequentially, modulators of the DEGs, including transcription factors (TFs) and microRNAs, were predicted by the ChIP Enrichment Analysis databases and WEB-based GEne SeT AnaLysis Toolkit system, respectively. In addition, the protein-protein interaction network of DEGs was constructed via the search tool for the retrieval of interacting genes and then the functional modules were further analyzed via the cluster-Maker package and The Biological Networks Gene Ontology package within the Cytoscape software. A total of 482 DEGs, including 279 upregulated and 203 downregulated DEGs, were screened out. DEGs were predominantly enriched in the pathways of fatty acid metabolism, cardiac muscle contraction and DNA replication. TFs, including SMAD4, in addition to microRNAs, including the microRNA-125 (miR-125) family, miR-331 and miR-24, may be the modulators of the DEGs in PO. In addition, the five largest modules were identified with TTN, L1G1, ACADM, UQCRC2 and TRIM63 as the hub proteins, and they were associated with the biological processes of muscle contraction, DNA replication initiation, lipid modification, generation of precursor metabolites and energy, and regulation of acetyl-CoA biosynthetic process, respectively. SMAD4, CACNG1 and TRIM63 are suggested to be important factors in the molecular mechanisms of PO, and miR-331 may be novel potential biomarker for PO.
Collapse
Affiliation(s)
- Yanqing Liu
- Department of Geriatric Medicine, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Yueqiu Wang
- Department of Joint Brach, Jining No. 2 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Nailong Yang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Suning Wu
- Department of Geriatric Medicine, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Yanhua Lv
- Department of Obstertrics and Gynecology, The Affiliated Hospital of Jining Medical College, Jining, Shandong 272000, P.R. China
| | - Lili Xu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
17
|
Song M, Zhao D, Wei S, Liu C, Liu Y, Wang B, Zhao W, Yang K, Yang Y, Wu H. The effect of electromagnetic fields on the proliferation and the osteogenic or adipogenic differentiation of mesenchymal stem cells modulated by dexamethasone. Bioelectromagnetics 2014; 35:479-90. [PMID: 25145543 DOI: 10.1002/bem.21867] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 05/17/2014] [Indexed: 01/29/2023]
Affiliation(s)
- Mingyu Song
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| | - Dongming Zhao
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| | - Sheng Wei
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| | - Chaoxu Liu
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| | - Yang Liu
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| | - Bo Wang
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| | - Wenchun Zhao
- Navy University of Engineering; Wuhan Hubei China
| | - Kaixiang Yang
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| | - Yong Yang
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| | - Hua Wu
- Department of Orthopedics; Tongji Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan Hubei China
| |
Collapse
|
18
|
Li J, Zhang N, Huang X, Xu J, Fernandes JC, Dai K, Zhang X. Dexamethasone shifts bone marrow stromal cells from osteoblasts to adipocytes by C/EBPalpha promoter methylation. Cell Death Dis 2013; 4:e832. [PMID: 24091675 PMCID: PMC3824658 DOI: 10.1038/cddis.2013.348] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 08/01/2013] [Accepted: 08/12/2013] [Indexed: 01/09/2023]
Abstract
Dexamethasone (Dex)-induced osteoporosis has been described as the most severe side effect in long-term glucocorticoid therapy. The decreased bone mass and the increased marrow fat suggest that Dex possibly shifts the differentiation of bone marrow stromal cells (BMSCs) to favor adipocyte over osteoblast, but the underlying mechanisms are still unknown. In this paper, we established a Dex-induced osteoporotic mouse model, and found that BMSCs from Dex-treated mice are more likely to differentiate into adipocyte than those from control mice, even under the induction of bone morphogenetic protein-2 (BMP2). We also discovered both in vitro and in vivo that the expression level of adipocyte regulator CCAAT/enhancer-binding protein alpha (C/EBPalpha) is significantly upregulated in Dex-induced osteoporotic BMSCs during osteoblastogenesis by a mechanism that involves inhibited DNA hypermethylation of its promoter. Knockdown of C/EBPalpha in Dex-induced osteoporotic cells rescues their differentiation potential, suggesting that Dex shifts BMSC differentiation by inhibiting C/EBPalpha promoter methylation and upregulating its expression level. We further found that the Wnt/beta-catenin pathway is involved in Dex-induced osteoporosis and C/EBPalpha promoter methylation, and its activation by LiCl rescues the effect of Dex on C/EBPalpha promoter methylation and osteoblast/adipocyte balance. This study revealed the C/EBPalpha promoter methylation mechanism and evaluated the function of Wnt/beta-catenin pathway in Dex-induced osteoporosis, providing a useful therapeutic target for this type of osteoporosis.
Collapse
Affiliation(s)
- J Li
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai Jiao Tong University School of Medicine (SJTUSM), China
| | | | | | | | | | | | | |
Collapse
|
19
|
The epithelial sodium channel is involved in dexamethasone-induced osteoblast differentiation and mineralization. Cell Biol Toxicol 2012; 28:279-89. [DOI: 10.1007/s10565-012-9222-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Accepted: 05/28/2012] [Indexed: 01/09/2023]
|